Inflammation and airway microbiota during cystic fibrosis pulmonary exacerbations

Edith T Zemanick, J Kirk Harris, Brandie D Wagner, Charles E Robertson, Scott D Sagel, Mark J Stevens, Frank J Accurso, Theresa A Laguna, Edith T Zemanick, J Kirk Harris, Brandie D Wagner, Charles E Robertson, Scott D Sagel, Mark J Stevens, Frank J Accurso, Theresa A Laguna

Abstract

Background: Pulmonary exacerbations (PEx), frequently associated with airway infection and inflammation, are the leading cause of morbidity in cystic fibrosis (CF). Molecular microbiologic approaches detect complex microbiota from CF airway samples taken during PEx. The relationship between airway microbiota, inflammation, and lung function during CF PEx is not well understood.

Objective: To determine the relationships between airway microbiota, inflammation, and lung function in CF subjects treated for PEx.

Methods: Expectorated sputum and blood were collected and lung function testing performed in CF subjects during early (0-3d.) and late treatment (>7d.) for PEx. Sputum was analyzed by culture, pyrosequencing of 16S rRNA amplicons, and quantitative PCR for total and specific bacteria. Sputum IL-8 and neutrophil elastase (NE); and circulating C-reactive protein (CRP) were measured.

Results: Thirty-seven sputum samples were collected from 21 CF subjects. At early treatment, lower diversity was associated with high relative abundance (RA) of Pseudomonas (r = -0.67, p<0.001), decreased FEV(1%) predicted (r = 0.49, p = 0.03) and increased CRP (r = -0.58, p = 0.01). In contrast to Pseudomonas, obligate and facultative anaerobic genera were associated with less inflammation and higher FEV₁. With treatment, Pseudomonas RA and P. aeruginosa by qPCR decreased while anaerobic genera showed marked variability in response. Change in RA of Prevotella was associated with more variability in FEV₁ response to treatment than Pseudomonas or Staphylococcus.

Conclusions: Anaerobes identified from sputum by sequencing are associated with less inflammation and higher lung function compared to Pseudomonas at early exacerbation. CF PEx treatment results in variable changes of anaerobic genera suggesting the need for larger studies particularly of patients without traditional CF pathogens.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Summary of most prevalent and…
Figure 1. Summary of most prevalent and top ranked (highest relative abundance) genera from early treatment CF sputum samples.
Prevalence of genera that are top ranked or detected in more than 25% of early-treatment sputum samples are shown. Grey bars show the percent of samples with a given bacterial genera detected by pyrosequencing. Black bars indicate the percent of samples in which the bacterial genera was the top ranked genus detected. (N = 21 early treatment sputum samples).
Figure 2. PCA plot of bacterial genera…
Figure 2. PCA plot of bacterial genera relative abundance and inflammatory markers at Early Treatment of PEx.
Colored dots represent individual subjects with subject identification number (SID) (n = 21). The length of the vectors represents the PCA loadings of the variables on the first two principal components, which explain 42% of the variability. Staphylococcus and Pseudomonas are both positively correlated with NE and CRP, while Veillonella, Granulicatella, Prevotella and Rothia are negatively correlated.. The first principal component (PC) was negatively correlated with FEV1% predicted indicating that sputum samples with higher inflammation and higher RA of Pseudomonas had lower FEV1 and those with lower inflammation and higher RA of Veillonella, Granulicatella or Prevotella had higher FEV1.
Figure 3. Cross-sectional relationship between airway microbiota,…
Figure 3. Cross-sectional relationship between airway microbiota, lung function and inflammation measured at Early Treatment of PEx.
(n = 21 subjects) Spearman’s rank correlation coefficients and 95% confidence intervals (bars) are shown, measuring the association between the relative abundance of each genera with FEV1% predicted, C-reactive protein (CRP) and sputum neutrophil elastase (NE).
Figure 4. Relationship between diversity, FEV 1…
Figure 4. Relationship between diversity, FEV1 and Pseudomonas RA.
Lower diversity is associated with lower FEV1 percent predicted and higher RA of Pseudomonas. Circle size corresponds to relative abundance of Pseudomonas demonstrating that samples with larger relative abundance of Pseudomonas have lower diversity, while samples with no or low relative abundance of Pseudomonas (small circles) generally have higher diversity.
Figure 5. Bacterial genera with the largest…
Figure 5. Bacterial genera with the largest magnitude of change in relative abundance by pyrosequencing with treatment of PEx.
The median values for samples where the genus was detected during early treatment are connected with a thick black line. Statistically significant and marginally significant differences are represented by **(p = 0.01) and *(p = 0.06), for Pseudomonas and Staphylococcus respectively. (N = 16 subjects).
Figure 6. Contribution of change in relative…
Figure 6. Contribution of change in relative abundance of the genera Staphylococcus (Staph), Pseudomonas (Pseudo), and Prevotella (Prev) and all three combined in explaining the variation in response to treatment measured by change in FEV1% predicted.
The height of the bars correspond to the R-squared values, the lines indicate the upper 95% confidence intervals. The black bar corresponds to a model with all three predictors while the grey bars contain one predictor; R-squared values should only be compared across models with the same number of predictors. *Indicates p-values less than 0.05.

References

    1. Gibson RL, Burns JL, Ramsey BW (2003) Pathophysiology and management of pulmonary infections in cystic fibrosis. Am J Respir Crit Care Med 168: 918–951.
    1. Amadori A, Antonelli A, Balteri I, Schreiber A, Bugiani M, et al. (2009) Recurrent exacerbations affect FEV(1) decline in adult patients with cystic fibrosis. Respir Med 103: 407–413.
    1. Liou TG, Adler FR, Fitzsimmons SC, Cahill BC, Hibbs JR, et al. (2001) Predictive 5-year survivorship model of cystic fibrosis. Am J Epidemiol 153: 345–352.
    1. Sanders DB, Bittner RC, Rosenfeld M, Redding GJ, Goss CH (2011) Pulmonary exacerbations are associated with subsequent FEV1 decline in both adults and children with cystic fibrosis. Pediatr Pulmonol 46: 393–400.
    1. Goss CH, Burns JL (2007) Exacerbations in cystic fibrosis. 1: Epidemiology and pathogenesis. Thorax 62: 360–367.
    1. Zemanick ET, Wagner BD, Harris JK, Wagener JS, Accurso FJ, et al. (2010) Pulmonary exacerbations in cystic fibrosis with negative bacterial cultures. Pediatr Pulmonol 45: 569–577.
    1. Sanders DB, Bittner RC, Rosenfeld M, Hoffman LR, Redding GJ, et al. (2010) Failure to recover to baseline pulmonary function after cystic fibrosis pulmonary exacerbation. Am J Respir Crit Care Med 182: 627–632.
    1. Sibley CD, Rabin H, Surette MG (2006) Cystic fibrosis: a polymicrobial infectious disease. Future Microbiol 1: 53–61.
    1. Rogers GB, Carroll MP, Serisier DJ, Hockey PM, Jones G, et al. (2004) Characterization of bacterial community diversity in cystic fibrosis lung infections by use of 16s ribosomal DNA terminal restriction fragment length polymorphism profiling. J Clin Microbiol 42: 5176–5183.
    1. Bittar F, Rolain JM (2010) Detection and accurate identification of new or emerging bacteria in cystic fibrosis patients. Clin Microbiol Infect 16: 809–820.
    1. Hamady M, Walker JJ, Harris JK, Gold NJ, Knight R (2008) Error-correcting barcoded primers for pyrosequencing hundreds of samples in multiplex. Nat Methods 5: 235–237.
    1. Harris JK, De Groote MA, Sagel SD, Zemanick ET, Kapsner R, et al. (2007) Molecular identification of bacteria in bronchoalveolar lavage fluid from children with cystic fibrosis. Proc Natl Acad Sci U S A 104: 20529–20533.
    1. Bittar F, Richet H, Dubus JC, Reynaud-Gaubert M, Stremler N, et al. (2008) Molecular detection of multiple emerging pathogens in sputa from cystic fibrosis patients. PLoS ONE 3: e2908.
    1. Tunney MM, Field TR, Moriarty TF, Patrick S, Doering G, et al. (2008) Detection of anaerobic bacteria in high numbers in sputum from patients with cystic fibrosis. Am J Respir Crit Care Med 177: 995–1001.
    1. Tunney MM, Klem ER, Fodor AA, Gilpin DF, Moriarty TF, et al. (2011) Use of culture and molecular analysis to determine the effect of antibiotic treatment on microbial community diversity and abundance during exacerbation in patients with cystic fibrosis. Thorax 66: 579–584.
    1. Worlitzsch D, Rintelen C, Bohm K, Wollschlager B, Merkel N, et al. (2009) Antibiotic-resistant obligate anaerobes during exacerbations of cystic fibrosis patients. Clin Microbiol Infect 15: 454–460.
    1. Sibley CD, Parkins MD, Rabin HR, Duan K, Norgaard JC, et al. (2008) A polymicrobial perspective of pulmonary infections exposes an enigmatic pathogen in cystic fibrosis patients. Proc Natl Acad Sci U S A 105: 15070–15075.
    1. Klepac-Ceraj V, Lemon KP, Martin TR, Allgaier M, Kembel SW, et al. (2010) Relationship between cystic fibrosis respiratory tract bacterial communities and age, genotype, antibiotics and Pseudomonas aeruginosa. Environ Microbiol 12: 1293–1303.
    1. Laguna TA, Wagner BD, Luckey HK, Mann SA, Sagel SD, et al. (2009) Sputum desmosine during hospital admission for pulmonary exacerbation in cystic fibrosis. Chest 136: 1561–1568.
    1. Laguna TA, Wagner BD, Starcher B, Luckey Tarro HK, Mann SA, et al. (2012) Urinary desmosine: A biomarker of structural lung injury during CF pulmonary exacerbation. Pediatr Pulmonol 47: 856–863.
    1. Zemanick ET, Harris JK, Wagner BD, Robertson CE, Sagel SD, et al.. (2011) Relationship between lung microbiome, lung function and inflammation during treatment of CF pulmonary exacerbation. Pediatric Pulmonol Supplement 34: 306.
    1. Collaco JM, Green DM, Cutting GR, Naughton KM, Mogayzel PJ Jr (2010) Location and duration of treatment of cystic fibrosis respiratory exacerbations do not affect outcomes. Am J Respir Crit Care Med 182: 1137–1143.
    1. VanDevanter DR, O’Riordan MA, Blumer JL, Konstan MW (2010) Assessing time to pulmonary function benefit following antibiotic treatment of acute cystic fibrosis exacerbations. Respir Res 11: 137.
    1. Miller MR, Hankinson J, Brusasco V, Burgos F, Casaburi R, et al. (2005) Standardisation of spirometry. Eur Respir J 26: 319–338.
    1. Burns JL, Emerson J, Stapp JR, Yim DL, Krzewinski J, et al. (1998) Microbiology of sputum from patients at cystic fibrosis centers in the United States. Clin Infect Dis 27: 158–163.
    1. Matsuda K, Tsuji H, Asahara T, Kado Y, Nomoto K (2007) Sensitive quantitative detection of commensal bacteria by rRNA-targeted reverse transcription-PCR. Appl Environ Microbiol 73: 32–39.
    1. Nadkarni MA, Caldon CE, Chhour KL, Fisher IP, Martin FE, et al. (2004) Carious dentine provides a habitat for a complex array of novel Prevotella-like bacteria. J Clin Microbiol 42: 5238–5244.
    1. Harris JK, Sahl JW, Castoe TA, Wagner BD, Pollock DD, et al. (2010) Comparison of normalization methods for construction of large, multiplex amplicon pools for next-generation sequencing. Appl Environ Microbiol 76: 3863–3868.
    1. Wheeler DL, Barrett T, Benson DA, Bryant SH, Canese K, et al. (2008) Database resources of the National Center for Biotechnology Information. Nucleic Acids Res 36: D13–D21.
    1. Frank DN (2009) BARCRAWL and BARTAB: software tools for the design and implementation of barcoded primers for highly multiplexed DNA sequencing. BMC Bioinformatics 10: 362.
    1. Nawrocki EP, Kolbe DL, Eddy SR (2009) Infernal 1.0: inference of RNA alignments. Bioinformatics 25: 1335–1337.
    1. Haas BJ, Gevers D, Earl AM, Feldgarden M, Ward DV, et al. (2011) Chimeric 16S rRNA sequence formation and detection in Sanger and 454-pyrosequenced PCR amplicons. Genome Res 21: 494–504.
    1. Cole JR, Wang Q, Cardenas E, Fish J, Chai B, et al. (2009) The Ribosomal Database Project: improved alignments and new tools for rRNA analysis. Nucleic Acids Res 37: D141–D145.
    1. Wang Q, Garrity GM, Tiedje JM, Cole JR (2007) Naive Bayesian classifier for rapid assignment of rRNA sequences into the new bacterial taxonomy. Appl Environ Microbiol 73: 5261–5267.
    1. Claesson MJ, O’Sullivan O, Wang Q, Nikkila J, Marchesi JR, et al. (2009) Comparative analysis of pyrosequencing and a phylogenetic microarray for exploring microbial community structures in the human distal intestine. PLoS ONE 4: e6669.
    1. Altschul SF, Gish W, Miller W, Myers EW, Lipman DJ (1990) Basic local alignment search tool. J Mol Biol 215: 403–410.
    1. Pruesse E, Quast C, Knittel K, Fuchs BM, Ludwig W, et al. (2007) SILVA: a comprehensive online resource for quality checked and aligned ribosomal RNA sequence data compatible with ARB. Nucleic Acids Res 35: 7188–7196.
    1. Magurran AE (2004) Measursing Biological Diversity. Malden, MA: Blackwell Publishing Company.
    1. Jost L (2006) Entropy and Diversity. Oikos 113: 363–375.
    1. Cohen J, Cohen P, West SG, Aiken LS (2003) Applied Multiple Regression/Correlation Analysis for the Behavioral Sciences. Mahwah, NJ: Lawrence Earlbaum Associates.
    1. Ordonez CL, Henig NR, Mayer-Hamblett N, Accurso FJ, Burns JL, et al. (2003) Inflammatory and microbiologic markers in induced sputum after intravenous antibiotics in cystic fibrosis. Am J Respir Crit Care Med 168: 1471–1475.
    1. Colombo C, Costantini D, Rocchi A, Cariani L, Garlaschi ML, et al. (2005) Cytokine levels in sputum of cystic fibrosis patients before and after antibiotic therapy. Pediatr Pulmonol 40: 15–21.
    1. Parkins MD, Sibley CD, Surette MG, Rabin HR (2008) The Streptococcus milleri group–an unrecognized cause of disease in cystic fibrosis: a case series and literature review. Pediatr Pulmonol 43: 490–497.
    1. Eribe ER, Olsen I (2008) Leptotrichia species in human infections. Anaerobe 14: 131–137.
    1. Zemanick ET, Wagner BD, Sagel SD, Stevens MJ, Accurso FJ, et al. (2010) Reliability of quantitative real-time PCR for bacterial detection in cystic fibrosis airway specimens. PLoS ONE 5: e15101.
    1. Ulrich M, Beer I, Braitmaier P, Dierkes M, Kummer F, et al. (2010) Relative contribution of Prevotella intermedia and Pseudomonas aeruginosa to lung pathology in airways of patients with cystic fibrosis. Thorax 65: 978–984.
    1. Rogers GB, Carroll MP, Serisier DJ, Hockey PM, Jones G, et al. (2006) Use of 16S rRNA gene profiling by terminal restriction fragment length polymorphism analysis to compare bacterial communities in sputum and mouthwash samples from patients with cystic fibrosis. J Clin Microbiol 44: 2601–2604.
    1. Goddard AF, Staudinger BJ, Dowd SE, Joshi-Datar A, Wolcott RD, et al. (2012) Direct sampling of cystic fibrosis lungs indicates that DNA-based analyses of upper-airway specimens can misrepresent lung microbiota. Proc Na,tl Acad Sci U S A 109: 13769–13774.
    1. Hilty M, Burke C, Pedro H, Cardenas P, Bush A, et al. (2010) Disordered microbial communities in asthmatic airways. PLoS ONE 5: e8578.
    1. Erb-Downward JR, Thompson DL, Han MK, Freeman CM, McCloskey L, et al. (2011) Analysis of the lung microbiome in the “healthy” smoker and in COPD. PLoS ONE 6: e16384.
    1. Cox MJ, Allgaier M, Taylor B, Baek MS, Huang YJ, et al. (2010) Airway microbiota and pathogen abundance in age-stratified cystic fibrosis patients. PLoS ONE 5: e11044.
    1. Zhao J, Schloss PD, Kalikin LM, Carmody LA, Foster BK, et al. (2012) Decade-long bacterial community dynamics in cystic fibrosis airways. Proc Natl Acad Sci U S A 109: 5809–5814.
    1. Fodor AA, Klem ER, Gilpin DF, Elborn JS, Boucher RC, et al. (2012) The adult cystic fibrosis airway microbiota is stable over time and infection type, and highly resilient to antibiotic treatment of exacerbations. PLoS ONE 7: e45001.
    1. Daniels TW, Rogers GB, Stressmann FA, van der Gast CJ, Bruce KD, et al. (2012) Impact of antibiotic treatment for pulmonary exacerbations on bacterial diversity in cystic fibrosis. J Cyst Fibros 12: 22–28.
    1. Rogers GB, Marsh P, Stressmann AF, Allen CE, Daniels TV, et al. (2010) The exclusion of dead bacterial cells is essential for accurate molecular analysis of clinical samples. Clin Microbiol Infect 16: 1656–1658.

Source: PubMed

3
订阅